Basic Principles and Pharmacodynamics

Published on 07/02/2015 by admin

Filed under Basic Science

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 10813 times

Chapter 1 Basic Principles and Pharmacodynamics

The term pharmacology is derived from the Greek words pharmakon, meaning drug, and logos, meaning rational discussion or study. Thus pharmacology is the rational discussion or study of drugs and their interactions with the body. Classically there are two major divisions of pharmacology: pharmacodynamics and pharmacokinetics. Pharmacodynamics is the study of actions of drugs on the body—what effects a drug has on the patient, including mechanisms of action, beneficial and adverse effects of the drug, and the drug’s clinical applications. Pharmacokinetics is the inverse: the study of actions of the body on drugs—the absorption, distribution, storage, and elimination of a drug. An emergent third division is pharmacogenomics: the study of how genetic makeup affects pharmacodynamics and pharmacokinetics and thus affects drug selection and application to individual patients.

There is no precise uniformly accepted definition for the term drug. However, it is commonly accepted that a drug is any exogenous non-nutritive substance that affects bodily function. Drugs may influence bodily functions via several general mechanisms, including physical interactions (e.g., antacids), by affecting enzymatic activity (e.g., increasing or decreasing), or by binding to molecular structures on or in the cell that affect cellular function (e.g., antihistamines).

Drug Nomenclature

Several names refer to the same drug, which can be a source of confusion for students and practitioners alike.

Drug-Receptor Interactions

Although some notable exceptions exist, a fundamental principle of pharmacology is that drugs must interact with a molecular target to exert an effect. Drug interaction with molecular targets is the initiating event in a multistep process that ultimately alters tissue function. For the purposes of current discussion, the target will be referred to as a receptor. An in-depth discussion of molecular targets and a description of these processes will be presented later in this chapter (see the discussion of molecular mechanisms of drug action). Let us first consider the relationship between drug binding to its target receptors and the ultimate response of the tissue.

At its most fundamental level, the interaction of drug and receptor follows the law of mass action. The law of mass action dictates that:

Factors Affecting Drug-Target Interactions

Two basic properties of the drug-receptor interaction contribute importantly to drug responses: the ability of the drug to bind to its receptor, and the ability of the drug to alter the activity of its receptor.

Drug Binding

At the molecular level, a number of factors contribute to the interaction between drug and receptor and control the strength, duration, and type of the drug-receptor interaction. Collectively these factors dictate the strength with which the drug forms a complex with its receptor, also known as the affinity:

It is important to recognize that, in most cases, binding of drug to target molecules involves weaker bonds. Accordingly, the drug-receptor complex is not static, but rather there is continuous association and dissociation of the drug with the receptor as long as drug is present. A measure of the relative ease with which the association and dissociation reactions occur is the equilibrium dissociation constant (KD). Each drug-receptor combination will have a characteristic KD value. Drugs with high affinity for a given receptor display a small value for KD, and vice versa. In Figure 1-1, A and B, Drug A has a higher affinity for the receptor than Drug B. KD also represents the concentration of drug needed to bind 50% of the total receptor population. These concepts are important in the study of basic pharmacologic data regarding different compounds with affinity for the same receptor. In general, drugs with lower KD values will require lower concentrations to achieve sufficient receptor occupancy to exert an effect.

Selectivity of Drug Responses

Another important and desirable facet of pharmacologic responses is selectivity of drug action, determined by drug molecules exhibiting preferential affinity for receptors, as follows:

It is important to note that selectivity of drug action is a key concept. Few drugs are entirely specific for one receptor. Rather, drugs exhibit selectivity toward different receptors based on their relative affinities. Thus, selectivity is also relative. As the concentration of a drug increases, the drug will combine with receptors for which it has lower affinity and may generate off-target effects.

Activation of the Molecular Target

The relationship between the drug-receptor binding event and the ultimate biologic effect is complex. Quite often in experimental settings, the KD (concentration causing 50% receptor occupancy) does not correspond to a 50% maximal response from the test tissue or organism. In fact, in many cases half-maximal tissue responses are obtained at drug concentrations below the KD, suggesting that amplification of drug response occurs. Amplification of drug responses is discussed in a later section. This observation suggests that other factors, in addition to affinity and receptor occupancy, determine the strength of response. Accordingly, an additional modifier termed intrinsic activity was proposed. Intrinsic activity indicates the ability of receptor-bound drug to activate the receptor and initiate downstream events, leading to an effect. Drugs are categorized based on their intrinsic activity at a given receptor:

Thus, the ultimate action of a drug will depend on both its affinity and its intrinsic activity. It is important to remember that affinity and intrinsic activity are distinct properties. A weak partial agonist, which by definition activates a receptor only minimally, may have very high affinity for a receptor. In this case the drug will be able to effectively compete for the receptor and will usually out-compete the endogenous agonist for receptor occupancy and inhibit the endogenous response.

Quantifying Drug-Target Interactions: Dose-Response Relationships

Ultimately, to make informed clinical decisions regarding drug treatment, it is necessary to understand the relationship between the amount of drug given and the anticipated effect in the patient. This relationship is described quantitatively by the dose-response curve. There are two basic types of dose-response curves—graded and quantal—and each provides useful information for therapeutic decisions.

Graded Dose-Response Curves

image Have a sigmoidal shape similar to the drug receptor occupancy curves shown in Figure 1-2, because the biologic response to a drug is determined by the interaction of a drug with a receptor or molecular target.

The ED50 and Emax are useful parameters to assess drugs. In Figure 1-2, A, Drug A is more potent than Drug B or Drug C, whereas Drugs B and C have equal potency. Potency is sometimes used incorrectly as a measure of therapeutic effectiveness. In fact, in most cases potency is secondary to Emax in drug selection. However, in situations in which the absorption of drug is very poor, such that only small quantities of the drug reach the target, potency can be a critical consideration. Drugs with higher Emax values have higher pharmacologic efficacy.

In Figure 1-2, A, Drug B has the greatest efficacy, followed by Drug C, whereas Drug A, despite being the most potent, has the least efficacy. Drug C is equipotent with Drug B but has less efficacy. Thus, potency and efficacy can vary independently. It is important not to confuse the pharmacologic usage of efficacy with the more general usage. Pharmacologic efficacy is a measure of the strength of effect produced by the maximum dose of drug. By definition, antagonists do not activate their receptors after binding and therefore have an intrinsic activity and efficacy of 0. Nevertheless, an antagonist may be very clinically “efficacious” or beneficial because it blocks activation of the receptor by endogenous agonist.

These variables can be useful in determining how much of a drug to administer. For example, knowledge of the ED50 concentration for blood pressure lowering can be used to determine the dose of antihypertensive agent to administer to achieve a certain magnitude of blood pressure reduction. However, the astute clinician recognizes that ED50 values are derived from the average of a great many patients and thus should be used only as initial guidelines. Because of interindividual variability, each patient may respond in ways that differ from the average. The second type of dose-response curves, quantal dose-response curves, provide an estimate of this variability.

Quantal Dose-Response Curves

Quantal dose-response curves do the following:

image Represent a cumulative frequency distribution for a given response.

image Can be plotted for therapeutic, toxic, and lethal effects to obtain:

Antagonism as a Mechanism of Drug Action

Although stimulation of molecular targets is a major mode of drug action, inhibition of stimulation by endogenous ligands is perhaps an even more important mechanism. In many disease states, excessive activation or sensitivity of endogenous physiologic pathways (e.g., bronchial hyperreactivity in asthma) occurs, and effective pharmacologic therapy acts to inhibit these pathways. The ways in which drugs act as antagonists can be classified into several general mechanisms, including the following:

Pharmacologic Antagonists

The majority of antagonists used as drug therapy are pharmacologic antagonists that act by directly interfering with an agonist’s ability to activate its molecular target. The antagonist prevents agonist binding or agonist activation of the receptor and inhibits the biologic effects generated by the agonist. The interaction between antagonist and agonist can take several forms, including competitive reversible, competitive irreversible, and noncompetitive antagonism.

image

image Competitive reversible antagonism (also called competitive surmountable antagonism)

image As the concentration of antagonist increases, the number of antagonist-receptor complexes increases and the number of agonist-receptor complexes decreases. Therefore the agonist effect decreases. Figure 1-3, A shows a graded dose-response curve for increasing concentrations of antagonist in the presence of a fixed concentration of agonist. The concentration of antagonist that reduces the agonist response to 50% of maximum is the IC50, one index for quantifying antagonist effectiveness. Note that IC50 values vary with agonist starting concentration.
image The agonist-antagonist relationship can also be depicted on agonist dose-response curves. Figure 1-3, B illustrates graded agonist dose-response curves in the absence (control) and presence of increasing doses of antagonist.

image Noncompetitive antagonism (also called allotropic or allosteric antagonism)

Molecular Mechanisms Mediating Drug Action

To this point we have considered the drug response as being elicited by agonist binding to and activating a receptor. In fact, there are many intervening steps between drug binding or receptor activation and the ultimate tissue response. In many cases this is because the drug is not able to interact directly with the cellular mechanisms eliciting the response. Instead, the drug must rely on intermediaries to relay (transduce) the drug signal to the cellular communication (second messenger signaling) and effector systems that ultimately cause the response. In addition, these transduction and signaling mechanisms are also integral in integrating convergent inputs to the cell and to modulation of drug responses by cells. In the preceding discussion, we used the general concept of receptor as the molecular target for drug action. In reality, there are many different types of molecular targets mediating drug responses. Knowledge of the different types of molecular targets and their associated transduction and signaling mechanisms aids in understanding drug action and the factors that modify drug responses.

Receptor Coupling and Transduction Mechanisms

In pharmacology, transduction refers to the conversion of the information contained in the drug molecule (e.g., size, shape) into a signal that can be recognized and acted on by the cell. This process of receptor coupling, or transduction, is critically important to generating the ultimate biologic response. Transduction events are also important mechanisms contributing to the sensitivity of tissues to most drugs. Only minute amounts of drug are generally necessary to initiate or inhibit a response, because transduction mechanisms greatly amplify the signal generated by the drug-target complex. Transduction generally involves a sequence of events that represent opportunities for interaction between different drug signals, for the cell to modulate the initial signal produced by the drug (feedback), and for future drug development. Indeed, many currently used drugs do not interact directly with endogenous substances or their receptors but rather interact with transduction events to cause their actions.

Extracellular Transduction Mechanisms

A number of dugs act outside of the cell to affect cellular function. Generally, these types of drugs act via the following:

Transmembrane Transduction Mechanisms

In many cases, the drug or endogenous ligand is a hydrophilic substance that cannot easily cross the plasma membrane of the cell and binds to receptors or other targets embedded in the plasma membrane. Accordingly, mechanisms are needed to transduce or relay the drug signal across the plasma membrane. It is possible to cluster these coupling and transduction mechanisms into several general groups (sometimes called superfamilies).

image Receptor-coupled enzymes. Receptor-coupled enzymes bypass the G protein coupling mechanism and link directly to cellular communication cascades. The receptor is directly coupled in some way to kinase enzymatic activity within the cell. Ligand binding stimulates the kinase enzymatic activity, which then initiates and amplifies intracellular signals and feedback responses by changing the phosphorylation status of cellular proteins. As shown in Figure 1-6, these mechanisms can be grouped into four general types that include receptors:

image Transmembrane ion channels. Transmembrane ion channels allow the passage of ions from one side of a membrane to another. Channels can exist in the open, closed, or inactive state, which represent different conformations of the channel protein. As shown in Figure 1-7, drugs may affect the function of these channels by directly opening or closing the channel (ligand gated channels), by influencing the voltage-dependent characteristics of the channels (voltage gated channels) and the amount of time the channel spends in a given state, or by generating second messengers that subsequently open or close the channel (second messenger gated). Common examples of functions governed by ion channels include the following:

Intracellular Transduction Mechanisms

A number of drugs bind to their primary site of action after being transported or diffusing into the intracellular space of the cell. Once inside the cell these receptors may be coupled to a number of transduction mechanisms, including transcriptional regulation, second messenger generation, and structural mechanisms.

Intracellular Receptors

Lipophilic drugs passively cross the cell membrane and thus do not require cell membrane receptors. As shown in Figure 1-8, one target for these drugs is an intracellular receptor that activates transcriptional pathways. In this mechanism, the agonist receptor complex diffuses to DNA, where it binds to DNA binding elements. Via this mechanism drugs act directly or through recruitment of coactivators or co-repressors, which increase or decrease transcription of RNA to ultimately change protein expression. This process is referred to as ligand gated transcriptional regulation. In many cases these drugs effect long-term changes by affecting gene transcription. Receptors using this coupling mechanism include:

Responses to these types of drug may be tissue dependent based on differential recruitment of coactivators or co-repressors. For example, selective estrogen receptor modulators (SERMs) acting on the same receptor may behave as an agonist in bone and as an antagonist in breast tissue (e.g., raloxifene).

Examples of ligand gated transcription regulation with clinical utility include:

Intracellular Enzymes

Some drugs directly target intracellular enzymes, such as phosphodiesterase (PDE), that control second messenger pathways (see Figure 1-8) and thereby alter the concentrations of intracellular signaling molecules, which then effects a cellular response. As greater understanding of intracellular signaling is achieved, it is likely that more drugs using this mode of action will be developed. Often there are multiple levels of intracellular signaling molecules downstream from the enzyme being targeted. A common example of the utility of this approach is PDE5 inhibition, to prevent the breakdown of cGMP, which results in increased vasodilation. This approach is useful in the treatment of erectile dysfunction because of the ability to somewhat selectively target blood vessels in the penis.

Second Messenger Systems

After formation of the drug-receptor complex and activation of a coupling mechanism (e.g., G proteins), the drug signal is transmitted to the final effector system of the cell. In many cases the transduction or coupling mechanism is linked to the final effector system via an intermediate cell signaling (second messenger) system. Drugs may also target enzymes or other processes regulating the concentrations of intracellular second messengers. This represents an important mode of drug action. In addition, it opens the possibility for synergistic or antagonist interactions among drugs that act at different sites in the same pathway. These interactions may enhance therapeutic effects or lead to adverse effects. The field of cell signaling is extremely dynamic, with new signaling molecules or new functions for established molecules discovered on a seemingly daily basis. Therefore, it is not possible to discuss the intricacies of all second messenger systems linked to clinically relevant drug actions. Nevertheless, several pathways serve as good illustrations of the involvement of cell signaling mechanisms as mediators of drug responses and as targets for future drug development. Figure 1-9 illustrates three of the best understood second messenger systems.

Cyclic Adenosine Monophosphate Pathway

Clinical Connection: Knowledge of coupling and second messenger systems can help in understanding drug action. Clinical use of β-adrenergic antagonists is associated with two apparently disparate effects. In the short term, β antagonists reduce cardiac function by blocking the formation of cAMP and signaling to the cardiac calcium channel. However, when used chronically in heart failure patients, β antagonists actually improve cardiac function, presumably via their long-term actions on gene transcription.

Phospholipase C, Inositol 1,4,5 Trisphosphate (IP3), Diacylglycerol (DAG)

Many more second messenger systems and signaling modalities exist and participate in drug responses. Improved understanding of this facet of pharmacology will point to greater opportunities for development of new drug targets.

Amplification of Drug Responses

Amplification is an important component of pharmacologic responses. A great deal of amplification occurs in pharmacologic pathways, such that only a minute quantity of drug (often in the picomolar or femtomolar range) is capable of eliciting biologic responses. In general, only minute concentrations of neurotransmitters, hormones, or exogenously administered drugs need reach the molecular target to initiate a biologic response. This exquisite sensitivity of tissues to drugs results in large part from amplification of the original signal provided by the drug molecule. Amplification can occur at several points in the drug-receptor coupling and signaling systems (Figure 1-10).

The net result is progressive amplification of the drug signal until the final effector system elicits a biologic response. Collectively, these mechanisms endow pharmacologic pathways with tremendous sensitivity, such that in general only minute amount of drug are necessary at the receptor to produce an effect.

Factors Modifying Drug Responses

The principles governing drug responses in overall terms were described earlier. It is important for clinicians to recognize that many of the parameters that have been discussed (e.g., ED50) were derived from population averages. However, in practice there is considerable variability in responsiveness to drugs among individuals. Drug responsiveness may also vary in the same patient over time or with disease progression. Therefore each patient will likely respond in a distinct manner. Variability in responsiveness may be an intrinsic feature of the patient, may be related to the disease process, or may occur in response to repeated administration of the drug. Multiple mechanisms may be involved, including:

Tachyphylaxis or desensitization refers to the relatively rapid (minutes, hours) changes in drug responsiveness caused by repeated drug administration.

Tolerance generally refers to reductions in responsiveness that occur over a longer time frame (days or longer) caused by prolonged drug administration.

Homologous desensitization or tolerance is specific to one receptor type or drug class.

Heterologous desensitization or tolerance affects many receptor types or drugs.